WO2009024620A2 - A process for isolation and purification of a target protein free of prion protein (prpsc) - Google Patents

A process for isolation and purification of a target protein free of prion protein (prpsc) Download PDF

Info

Publication number
WO2009024620A2
WO2009024620A2 PCT/EP2008/061068 EP2008061068W WO2009024620A2 WO 2009024620 A2 WO2009024620 A2 WO 2009024620A2 EP 2008061068 W EP2008061068 W EP 2008061068W WO 2009024620 A2 WO2009024620 A2 WO 2009024620A2
Authority
WO
WIPO (PCT)
Prior art keywords
protein
employing
buffer
wash buffer
buffer containing
Prior art date
Application number
PCT/EP2008/061068
Other languages
English (en)
French (fr)
Other versions
WO2009024620A3 (en
Inventor
Gustav Gilljam
Mats Jernberg
Stefan Winge
Andrea Neisser-Svae
Original Assignee
Octapharma Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP08787446.7A priority Critical patent/EP2190486B1/de
Priority to MX2010001919A priority patent/MX2010001919A/es
Priority to BRPI0815758-8A priority patent/BRPI0815758B1/pt
Priority to ES08787446T priority patent/ES2572352T3/es
Application filed by Octapharma Ag filed Critical Octapharma Ag
Priority to DK08787446.7T priority patent/DK2190486T3/en
Priority to US12/733,306 priority patent/US9296799B2/en
Priority to RU2010110805/10A priority patent/RU2491292C2/ru
Priority to CA2696865A priority patent/CA2696865C/en
Priority to JP2010521447A priority patent/JP5797404B2/ja
Priority to AU2008290482A priority patent/AU2008290482B2/en
Priority to CN200880104009XA priority patent/CN101842121B/zh
Publication of WO2009024620A2 publication Critical patent/WO2009024620A2/en
Publication of WO2009024620A3 publication Critical patent/WO2009024620A3/en
Priority to IL203573A priority patent/IL203573A/en
Priority to US14/982,148 priority patent/US20160152661A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/22Affinity chromatography or related techniques based upon selective absorption processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/38Selective adsorption, e.g. chromatography characterised by the separation mechanism involving specific interaction not covered by one or more of groups B01D15/265 - B01D15/36
    • B01D15/3804Affinity chromatography
    • B01D15/3809Affinity chromatography of the antigen-antibody type, e.g. protein A, G, L chromatography
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/38Selective adsorption, e.g. chromatography characterised by the separation mechanism involving specific interaction not covered by one or more of groups B01D15/265 - B01D15/36
    • B01D15/3804Affinity chromatography
    • B01D15/3828Ligand exchange chromatography, e.g. complexation, chelation or metal interaction chromatography
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/42Selective adsorption, e.g. chromatography characterised by the development mode, e.g. by displacement or by elution
    • B01D15/424Elution mode
    • B01D15/426Specific type of solvent
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M47/00Means for after-treatment of the produced biomass or of the fermentation or metabolic products, e.g. storage of biomass
    • C12M47/12Purification
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/34Size selective separation, e.g. size exclusion chromatography, gel filtration, permeation
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/36Selective adsorption, e.g. chromatography characterised by the separation mechanism involving ionic interaction
    • B01D15/361Ion-exchange
    • B01D15/362Cation-exchange
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/38Selective adsorption, e.g. chromatography characterised by the separation mechanism involving specific interaction not covered by one or more of groups B01D15/265 - B01D15/36
    • B01D15/3847Multimodal interactions
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J39/00Cation exchange; Use of material as cation exchangers; Treatment of material for improving the cation exchange properties
    • B01J39/26Cation exchangers for chromatographic processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J41/00Anion exchange; Use of material as anion exchangers; Treatment of material for improving the anion exchange properties
    • B01J41/20Anion exchangers for chromatographic processes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/165Extraction; Separation; Purification by chromatography mixed-mode chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/18Ion-exchange chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention pertains to a process for isolation and purification of a target protein which is free of the disease associated protein form PrP 50 .
  • PrP sc removal appears reproducible, incomplete understanding of the removal mechanism raises questions, such as how to (a) determine the maximum capacity of chromatographic support to bind TSE agents, (b) ensure efficient sanitizing procedures of recycled gels and (c) guarantee consistent PrP sc removal over production cycles (Thyer J, Prion- removal capacity of chromatographic and ethanol precipitation steps used in the production of albumin and immunoglobulins; Vox Sang. 2006 Nov; 91(4) : 292-300).
  • WO-A-98/0041 discloses removal of a prion from other proteins, e.g. haemoglobin, by ion exchange chromatography.
  • the preparation of the ion exchange chromatographic medium reveal silica gel derivatizesed with (g- glycidoxypropyl)trimethoxysilane and dimethanolamine to obtain a (uniform) surface of quaternary ammonium groups.
  • WO- A- 03/ 105911 discloses a cleaning method of human plasma by conventional means of ion exchange chromatography using a salt gradient for elution.
  • WO-A-94/08686 discloses a process for carrying out in a consecutive fashion different notes of chromatographic separation in a liquid chromatography column using a single separation medium.
  • D. B. Brimacombe et al. in Biochem. J. (1999) 342,605-613 discloses the purification of recPrP by two successive chromatographic steps.
  • the first step is a cation-exchange chromatography (150-650 NaCI-gradient) performed on S-Sepharose.
  • the pooled eluates of interest were subjected to a second chromatographic step (zink charged chelating sepharose; 0-10OmM imidazole gradient).
  • P. R. Foster et al. Vox Sanguinis 2000; 78:86-95 discloses the removal of prion protein in the manufacture of plasma products by many steps.
  • This method comprises 4 ion-exchange chromatographies (Steps 2, 11, 13 and 15) and one affinity chromatography (immobilised heparin on sepharose-FF; Step 12) performed in different columns on different chromatographic gels.
  • One object of the invention was to provide a chromatographic process which removes PrP sc during fractionation processes of sources being potentially contaminated by PrP sc , such as biologically derived sources.
  • the process should avoid the drawbacks of prior art.
  • Another object was to design a process which would render the purification process reliable and allowing regeneration of the chromatographic supports.
  • Still another object of the invention was to provide prion depleted fraction of proteins.
  • a process for isolation and purification of a target protein by chromatography wherein the chromatography removes or depletes prions (PrP sc ) is provided comprising the steps of
  • the process of the invention provides a significant improvement because the chromatography resin binds the PrP sc less strong enabling removal of the PrP sc from the chromatography resin before the target protein is eluted.
  • isolation and purification means in particular processes which are used to at least enrich any protein-like substances desired or processes which deplete unwanted substances. According to the invention it would be advantageous to yield the desired product as pure as possible.
  • target protein means a protein of interest which should be isolated and/or purified free of PrP sc .
  • the target protein can also be a still mixture of proteins if so desired e.g. a mixture of different factors having a biological effect when working in an ensemble.
  • Capto ® MMC and Capto ® Adhere from the company GE Healthcare, or MEP, HEA or PPA Hypercel ® resins from the company PALL.
  • the ligands of these multimodal resins can be differently designed; examples of different ligands are the following
  • the present invention provides a solid substrate that is an effective adsorbent for use in separating and isolating a variety of biological substances.
  • the solid substrate of this invention may be used, for example, in preparative techniques, such as column chromatography, and in analytical devices, such as biochips.
  • One advantage of the present solid substrate described herein is its high selectivity and specificity for biological substances such as immunoglobulins, together with the avoidance of costly and often detrimental cleaning processes required for prior art substrates.
  • a second advantage is that the solid substrate of this invention is almost ideally suited for use with biological samples at physiological pH and ionic strength, thereby obviating the need for pH adjustment and the addition of lyotropic salts as prescribed in the prior art.
  • As third advantage may be regarded the high capacity of the present substrates, which, in view the low cost of reagents employed to prepare them, presents significant economic gains over the use of specialized prior art adsorbents.
  • the solid substrates of this invention comprise a solid support and a ligand attached to the solid support.
  • the ligand comprises a cyclic group which can be a monocyclic group or a polycyclic group, and a linking group that optionally comprises a sulphur atom.
  • the ligand that attracts analytes through a mixed mode action is attached to a solid support.
  • the ligand comprises a cyclic group, which can be a monocyclic or polycyclic group that is tethered to the solid support and that is substituted by a sulphate, sulphonate, phosphate, or phosphonate group.
  • This monocyclic or polycyclic group can be an aromatic group, which, as defined here, is a cyclic hydrocarbon containing only unsaturated carbon-carbon bonds to give an aromatic system. While any aromatic group, in principle, may be employed in the present invention, a suitable aromatic group typically comprises one, two, or three aromatic rings. Thus, illustrative aromatic groups are phenyl and its substituted derivatives, such as tolyl and xylyl. Bicyclic aromatic groups comprise fused individual rings, and include but are not limited to naphthyl. Polycyclic aromatic groups include anthracenyl and phenanthrenyl, and groups such as acenanaphthylenyl that contain fused rings of different sizes. If an aromatic group is selected, it is preferred although not essential that the group be fused to a heterocyclic or heteroaromatic group, as described below.
  • a “heterocycle” is a saturated to partially saturated ring containing at least one hetero atom.
  • a “heteroaromatic group” is an aromatic group in which at least one carbon atom is substituted by a heteroatom.
  • the hetero atom preferably is N, S, or O. It also is preferable that the heterocyclic or heteroaromatic group is a five- or six-member ring, as reagents that comprise these groups are readily and inexpensively obtained from commercial sources.
  • heterocyclic or heteroaromatic group is one that establishes or contributes to the "thiophilic" character of the solid substrate, and is thus one that contains at least one S atom.
  • preferable heterocyclic or heteroaromatic groups may comprise at least one N atom, or combinations of S and N atoms.
  • exemplary heterocyclic or heteroaromatic groups include thiazoline, thiazolidone, imidazole, imidazoline, thiazole, triazoles, tetrazole, thiadiazole, imidazole, pyridine, and morpholine.
  • a suitable heterocyclic or heteroaromatic group is fused to an aromatic group, as deseribed above.
  • benzimidazole and benzothiazole are the readily available candidates, yielding superior solid substrates.
  • the monocyclic or polycyclic group is substituted with a sulphate, sulphonate, phosphate, or phosphonate group.
  • These groups are sufficiently acidic to exist as charged moieties within a large pH range, e.g., from about 2 to about 12.
  • the solid support is ideally suited to adsorb biological substances such as immunoglobulins at physiological ionic strength and pH.
  • substituted refers to the direct or indirect attachment of a sulphate, sulphonate, phosphate, or phosphonate group to the monocyclic or polycyclic group.
  • spacer group which is a Ci -6 straight or branched alkylene group.
  • the alkylene group is optionally interrupted by one or more bivalent moieties that include but are not limited to -C(O)NH-, NHC(O)-, -0-, -5-, -5(0)-, -5(O) 2 -, -NH-, - C(O)O-, and -OC(O)-.
  • illustrative spacer groups include -CH 2 -, -CH 2 CH 2 -, -CH 2 -O-CH 2 -, and -CH 2 C(O)NHCH 2 CH 2 -.
  • the monocyclic or polycyclic group is tethered to the solid support by a linking group, which comprises a mercapto, ether, or amino containing moiety.
  • a linking group which comprises a mercapto, ether, or amino containing moiety.
  • the linking group is hydrophobic, thereby conferring hydrophobic character to the solid support at a pH where binding of a biological substance occurs through both electrostatic and hydrophobic interactions.
  • Hydrophobic moieties include but are not limited to straight and branched Ci -6 alkylene groups, C 2-6 alkenylene groups, and C 2-6 alkynylene groups. Particularly useful moieties are ethylene and propylene. Other hydrophobic moieties comprise an aromatic group, as described above, to form, for example, phenethylene.
  • the linking group preferably contains a mercapto moiety.
  • the linking group confers hydrophobic and thiophilic characters to the solid substrate.
  • One preferred mercapto-containing linking group is represented by the formula:
  • the hydrophobicity of the linking group can be readily tailored by introducing polar substituents, such as hydroxyl, a halide, or nitro; by oxidizing a mercapto moiety by known methods; by incorporating ether or amino moieties into the linking group; or combinations thereof.
  • polar substituents such as hydroxyl, a halide, or nitro
  • oxidizing a mercapto moiety by known methods
  • ether or amino moieties into the linking group
  • one such mercapto- containing linking group that is readily accessed is represented by the formula :
  • an illustrative amino-containing linking group is represented by the formula:
  • the solid substrates comprised of amino-containing linking groups, or those containing oxidized mercapto moieties also comprise monocyclic er polycyclic groups that comprise at least one S atom.
  • the solid substrate is able to retain some thiophilic character.
  • the linking group itself comprises a polysaccharide such as hydroxy-ethyl-cellulose, starch, amylose, or agarose.
  • a preferred polysaccharide in this context is dextran.
  • the solid support is modified with a polysaccharide, which can be derivatized with a linking group as described below.
  • the solid substrate of this invention operates via "mixed-modes" of interaction between the solid substrate and a biological substance.
  • the aforementioned monocyclic and polycyclic groups have a pK-value below 4 and, hence, are negatively charged within the pH ranges of use as described above.
  • a biological substance such as an immunoglobulin, is contacted with the solid substrate between about pH 4 and pH 6, in which range the biological substance bears a net positive or neutral charge. In this pH range, the biological substance binds to the solid substrate through one or more types of interactions with the mono or polycyclic groups. The interactions include coulombic attractions and mild hydrophobic associations.
  • the solid support is of the form typically used for chromatography media, that is, a bead or particle. These beads or particles are derivatized with the mixed mode ligand. The beads or particles form a chromatography medium that one can use to pack the column.
  • the solid support takes the form of a chip, that is, a solid support having a generally planar surface to which the mixed mode ligand can be attached, covalently or otherwise. Chips that are adapted to engage a probe interface of a detection device are also called "probes.”
  • the solid substrate first comprises a solid support, which may comprise an organic material.
  • organic materials are polysaccharides, such as cellulose, starch, agar, agarose, and dextran.
  • Hydrophilic synthetic polymers are contemplated, including substituted or unsubstituted polyacrylamides, polymethacrylamides, polyacrylates, polymethacrylates, polyvinyl hydrophilic polymers, polystyrene, polysulfone, and copolymers or styrene and divinylbenzene.
  • inorganic materials may be used as the solid support material.
  • Such inorganic materials include but are not limited to porous mineral materials, such as silica; hydrogel containing silica, zirconia, titania, alumina; and other ceramic materials.
  • mixtures of these materials, or composite materials formed by copolymerization of or by an interpenetrated network of two materials such as those disclosed in US-A-5,268,097, US-A-5,234,991, and US-A-5,075,371.
  • the solid support may be in the form of beads or irregular particles of about 0.1 mm to about 1,000 mm in diameter.
  • the solid support can be fashioned into fibres, membranes, or sponge-like materials permeated with holes in the micron to multi-millimetre sizes.
  • the monocyclic or polycyclic groups described above are chemically immobilized on the solid support by forming covalent bonds between the solid support and the linking group, and between the linking group and monocyclic or polycyclic groups.
  • the solid support is first treated with a bifunctional reagent which serves to introduce onto the solid support reactive groups that form part or the entire linking group.
  • a bifunctional reagent which serves to introduce onto the solid support reactive groups that form part or the entire linking group.
  • a hydroxide source for example, prior to reaction with a bifunctional reagent.
  • the bifunctional reagent is capable of reacting both with the solid support and .with reagents that contain the monocyclic or polycyclic groups.
  • Illustrative bifunctional reagents which contain the same or different functional groups, include but are not limited to epichlorhydrin, epibromhydrin, dibromo- and dichloropropanol, dibromobutane, ethylene glycol diglycidylether, butanediol diglycidylether, divinyl sulfone, allylglycidylether, and allyl bromide.
  • the solid support is then washed extensively with one or more solvents to remove unreacted bifunctional reagent, reaction byproducts, or both.
  • a typical solvent used in this regard is water.
  • the monocyclic or polycyclic groups then are introduced by way of reagents that contain such groups substituted with mercapto, hydroxyl, or amino groups.
  • reagents that contain such groups substituted with mercapto, hydroxyl, or amino groups.
  • Such reagents react with functional groups presented by the functionalized solid support as described above.
  • a bifunctional reagent with a monocyclic or polycyclic reagent is guided by well-known chemistries.
  • solid supports that are functionalized with epoxides may undergo reactions with mercapto, hydroxy, or amino-containing reagents to furnish a substrate with ethylene- containing linking groups.
  • the alkene groups can be further brominated to furnish suitably reactive brome derivatives.
  • the concentration of immobilized monocyclic or polycyclic group can vary between a fraction of a micromole to several hundred micromoles per millilitre of solid support, depending upon the concentration of bifunctional reagent used to make the solid support.
  • PrP sc removal resin which mainly does not bind to the PrP sc one being that it is possible to extensively wash the resin with different buffer compositions before eluting the product, which makes it possible to at least reduce the number of PrP sc of even different biochemical composition to a very low level or remove PrP sc from the composition.
  • Detergents, alcohols and amino acids are also examples, which can be added to the washing buffers to achieve an optimal purity before eluting the product. It is significant more difficult to perform similar washing step to other types of "standard” chromatography media like for examples different types of ion exchange resins where the PrP sc binds to the resin. Even if the binding affinity is significant higher compared to product there will always be a risk that PrP sc to some degree will co-elute from the resin together with the product. Therefore there is a big advantage of using a resin in which the product "multimodal" binds to the resin whereas PrP sc have a relatively low affinity to the resin, which makes it possible to apply appropriate washing steps before eluting the product.
  • the target protein is eluted after PrP sc elution e.g. by amending the ionic strength of the elution buffer by increasing or decreasing the ionic strength, adding alcohols to the elution buffer - in particular in aqueous solution - such as mono- or dihyroxyalkanols, e.g. lower aliphatic alcohols such as methanol, ethanol, propanol, and/or amending the pH-value of the elution buffer by increasing or decreasing the pH.
  • aqueous solution - such as mono- or dihyroxyalkanols, e.g. lower aliphatic alcohols such as methanol, ethanol, propanol, and/or amending the pH-value of the elution buffer by increasing or decreasing the pH.
  • the invention uses increased ionic stength and increased ethylene glycol amount.
  • the elution of the target protein depends on the biochemical property of the target protein. For example it is possible to use the co-enzyme of the target protein or other substances with a high recognition towards the tertial structure of the target protein, e.g. antithrombin as target protein which elutes with an increased concentration of heparin.
  • the prion protein reduction value in the protein fraction comprising the target protein is > 1 to 4 Ig(IO), as calculated from the amount which was initially applied to the resin.
  • the prion analytical value in the protein fraction of interest is below detection limit of the prion Western blot assay.
  • the chromatographic conditions comprise at least two of the following steps: i) Employing a loading and equilibration buffer containing a solvent and/or a non-ionic detergent; ii) Employing a wash buffer without a solvent and / or non-ionic detergent; iii) Employing a wash buffer containing an alcohol and /or an amino acid; iv) Employing a wash buffer containing a high salt concentration; v) Employing a wash buffer containing a low salt concentration; vi) Employing a buffer containing a combination of alcohol and high salt concentration.
  • the chromatographic conditions comprise at least two of the following steps: i) Employing a loading and an equilibration buffer containing a solvent and or a non-ionic detergent; ii) Employing a first wash buffer which is a buffer without a solvent and a non-ionic detergent; iii) Employing a second wash buffer containing an alcohol and an amino acid; iv) Employing a third wash buffer containing high salt concentration; v) Employing a fourth wash buffer containing low salt concentration; vi) Employing an elution buffer containing a combination of an alcohol and high salt concentration.
  • the buffers employed are as follows i) Loading and equilibration buffer contain tri-n-butylphosphate and/or Triton x-100 in a concentration ranging of from about 0.1 to about 10% (w/w); ii) The second wash buffer contains ethylene glycol and/or lysine/arginine ranging of from about 5 to about 30 % (w/w) of ethylene glycol and of from 0.2 to about 1.5 M lysine/arginine; iii) The third wash buffer contains sodium chloride in a concentration ranging of from about 0.5 to about 4 M, in particular of from about 0.5 to about 1.5 M; iv) The fourth wash buffer contains sodium chloride in concentration ranging of from about 0.01 to about 0.2, in particular 0.01 to about
  • the elution buffer contains ethylene glycol and/or sodium chloride ranging in concentration of from about 25 to about 75 % (w/w), in particular of from about 25 to about 50 % of ethylene glycol and of from about 0.5 to about 4 M NaCI.
  • the chromatographic conditions comprise at least two of the following steps: i) Loading and equilibration buffer contain tri-n-bytylphosphate and/or Triton x-100 in a concentration ranging from 0.3 - 5% (w/w); ii) Washing with >10 column volumes of a second wash buffer containing ethylene glycol and/or lysine/arginine ranging from 10- 25 % (w/w) of EG and 0.3-1.0 M lysine/arginine; iii) Washing with >10 column volumes of a third wash buffer containing sodium chloride in a concentration ranging from 0.8-1.5 M; iv) Washing with > 10 column volumes of a fourth wash buffer containing sodium chloride in concentration ranging from 0.03-0.15 M; v) The elution buffer contains ethylene glycol and/or sodium chloride ranging in concentration from 35-65 % (w/w) for EG and 0.8-3.0 Na
  • the buffers employed are as follows i) Loading and equilibration buffer contain tri-n-bytylphosphate and/or Triton x-100 in a concentration ranging from 0.8 - 1.2 % (w/w); ii) Washing with >20 column volumes of a second wash buffer containing ethylene glycol and/or lysine/arginine ranging from 18- 22 % (w/w) of EG and 0.4-0.6 M lysine/arginine; iii) Washing with >20 column volumes of a third wash buffer containing sodium chloride in a concentration ranging from 0.8-1.2 M; iv) Washing with >20 column volumes of a fourth wash buffer containing sodium chloride in a concentration ranging from 0.08- 0.12 M; v) The elution buffer contains ethylene glycol and/or sodium chloride ranging in concentration from 45-55 % (w/w) for EG and 1.3-1.7
  • washing buffers of different types This increases the possibility that prions of different types and which binds due to to different interactions to the resin or the target protein, can be removed. Also by increasing the amount of respectively washing buffer (i.e one column volume is equal to the volume of the resin)the security of any remaining prions "slowacting" on the buffer applied, can be increased.
  • the multimodal chromatographic material may contain i) a positive charged N-Benzyl-N-methyl ethanolamine ligand; ii) a negatively charged 2- (benzoylamino) butanoic acid ligand; iii) a phenylpropyl ligand; iv) a N-hexyl ligand; v) a 4-Mercapto-Ethyl-Pyridine ligand; vi) a 3-[ ⁇ 3 ⁇ methyl-5-((tetrahydrofuran-2-ylmethyl)amino)- phenyl ⁇ amino]benzoic acid ligand.
  • Subject matter of the present invention is also a prion protein depleted fraction of a protein isolated from potentially infectious protein containing sources.
  • the fraction contains pharmaceutically applicable proteins obtainable according to the method of the invention.
  • protein fractions comprising plasma proteins, peptide hormones, growth factors, cytokines and polyclonal immunoglobulins proteins, plasma proteins selected from human and animal blood clotting factors including fibrinogen, prothrombin, thrombin, prothrombin complex, FX, FXa, FIX, FIXa, FVII, FVIIa, FXI, FXIa, FXII, FXIIa, FXIII and FXIIIa, von Willebrandt factor, transport proteins including albumin, transferrin, ceruloplasmin, haptoglobin, hemoglobulin and hemopexin, protease inhibitors including ⁇ -antithrombin, ⁇ -antithrombin, cc2-macroglobulin, Cl-inhibitor, tissue factor pathway inhibitor (TFPI), heparin cofactor II, protein C inhibitor (PAI-3), Protein C and Protein S, ⁇ -1 esterase inhibitor proteins, ⁇ -1 antitrypsin, antiangion
  • Tricorn column (GE Healthcare, Sweden, cross sectional area: 0.2cm 2 , diameter 0.5 cm) was packed with Capto MMC resin (GE Healthcare Cat. No. 17-5317-10, lot No. 308581), 9 cm bed height, Column volume: 1.8 ml.
  • Buffer compositions * Buffer 1 (Equilibration buffer with S/D added)
  • Table 1 Outlining the approximate amounts of buffer applied, flow rates expressed as ml/min as well as cm/hour. Time required for each buffer step and contact time with the gel for the protein solution is also shown.
  • Elution buffer 1.5 M NaCI 7 12 0,20 61 62 8,8 MMC resin, packed in a Tricorn column with a bed height of approx. 9 cm.
  • the chromatography step was monitored for conductivity and at 280nm.
  • the protein load was approximately 3mg related to 1 ml of resin.
  • the column was properly equilibrated with equilibration buffer containing S/D chemicals until a stable base line was obtained.
  • the starting material was added S/D chemicals at a ratio of 14 g S/D stock per kg to obtain the same concentration as the equilibration buffer, this was stirred for at least 10 minutes before application of the protein solution to the column.
  • Fractions of the following buffers where collected and analysed for total protein (and prions in the PrP sc spiking experiments)
  • the chromatography profile measured at an absorbance of 280 nm can be seen in appendix 2 :
  • Buffer 1 High non-ionic detergent Buffer; 0.3 M NaCI, 0.01 M CaCI 2 , 0.01 M L-Histidin, 1% w/w Triton X-100, 0.3% w/w TNBP, pH : 7.0
  • Buffer 2 Low non-ionic detergent Buffer; 0.3 M NaCI, 0.01 M CaCI 2 ,
  • Buffer 3 Amino acid / Alcohol Buffer; 0.3 M NaCI, 0.01 M CaCI 2 , 0.01 M L-Histidin, 0.02% (w/w) Tween 80, 0.5 M L-Lysin monochlorid, 20% (w/w) Ethylene glycol, pH 6.5
  • Buffer 4 High salt Buffer; 1.0 M NaCI, 0.05 M CaCI 2 , 0.05 M L-Histidin,
  • Buffer 5 Low salt Buffer; 0.1 M NaCI, 0.01 M CaCI 2 , 0.01 M L-Histidin, 0.02% (w/w) Tween 80 pH : 6.5 )
  • na Not analysed due to interference of buffer with total protein analytical method
  • Example 1 To be able to determine the prion protein removal of the chromatography procedure described in example 1, a prion spiking experiment was performed. The same column, resin, buffers and starting material as in Example 1 was used.
  • Triton X-100 1.955 g of Triton X-100 were mixed with 0.582 g of TnBP (target ratio: 10 parts + 3 parts, determination per weight) and stirred for 36 min.
  • 0.665 g of the S/D reagent were then immediately added to the remaining 47.72 g of spiked start material (target ratio: 14 g S/D-reagent per kg of spiked start material) and stirred for 31 min.
  • the temperature of the start material was checked to be 24.5°C in the beginning and 23.7°C at the end of the stirring phase (target range: 18-25°C).
  • Buffer 1 Equilibration buffer with S/D
  • the column was flushed with 10.0 CV of Buffer 2 (Equilibration Buffer without S/D) at a flow rate of 0.8 ml/min (target: 10 CV at 1.0 ml/min). Collection of the flow through started when the UV signal began to rise and was continued until the absorbance started to drop. The weight of the flow through fraction was determined (actual weight: 48.23 g), a 16 ml aliquot removed, aliquoted and stored at ⁇ -60 0 C (sample flow through- FT). Wash fraction 1 was collected during flushing with Buffer 2. The actual weight of this fraction was determined to be 12.75 g and a 12 ml aliquot was removed and stored at ⁇ -60 0 C (sample washl-Wl).
  • Buffer 2 Equilibration Buffer without S/D
  • wash fraction 3 was collected. An actual weight of 18.48 g was determined, a 16 ml aliquot was then removed, aliquotted and stored at ⁇ -60 0 C (sample wash3-W3).
  • wash fraction 4 was collected. The actual weight of this fraction was determined to be 12.22 g. A 11.5 ml aliquot was removed and stored at ⁇ -60 0 C (sample wash4-W4).
  • the product was then eluted with 8.3 CV of Buffer 6 (Elution Buffer), applying a flow rate of 0.2 ml/min (target: 7 CV at 0.2 ml/min). Collection of the eluate was carried out during the whole period of flushing the column with Buffer 6. The actual weight of the eluate fraction was determined to be 13.54 g, a 12.5 ml aliquot was removed and stored at ⁇ -60 0 C (sample eluate-E). During regeneration of the column with 9.4 CV of Buffer 7 (Regeneration Buffer) at a flow rate of 0.6 ml/min (target: 20 CV at 0.6 ml/min), the regeneration fraction was collected. An actual weight of 17.97 ml was determined, a 16 ml aliquot removed and stored at ⁇ -60 0 C (sample regenaration-Reg).
  • Protein determination according to Bradford is based on the observation that the absorbance maximum for an acidic solution of Coomassie Brilliant Blue G- 250 shifts from 465 nm to 595 nm when binding to protein occurs. Both hydrophobic and ionic interactions stabilize the anionic form of the dye, causing a visible colour change.
  • the assay is useful since the extinction coefficient of a dye-albumin complex solution is constant over a 10-fold concentration range. See for further information also Bradford, MM. A rapid and sensitive for the quantitation of microgram quantitites of protein utilizing the principle of protein-dye binding. Analytical Biochemistry 72: 248-254. 1976. Western Blot Assay for the Detection of PrP Sc
  • the Western blot assay is a semi-quantitative determination of proteinase K resistant Scrapie associated prion protein (PrP Sc ).
  • the Western blot assay was performed as described by DC Lee et al., Journal of Virological Methods 2000;84:77-89.
PCT/EP2008/061068 2007-08-23 2008-08-25 A process for isolation and purification of a target protein free of prion protein (prpsc) WO2009024620A2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
US12/733,306 US9296799B2 (en) 2007-08-23 2008-08-25 Process for isolation and purification of a target protein free of prion protein (PrPSC)
BRPI0815758-8A BRPI0815758B1 (pt) 2007-08-23 2008-08-25 PROCESSO PARA O ISOLAMENTO E A PURIFICAÇÃO DE UMA PROTEÍNA-ALVO LIVRE DE PROTEÍNA PRÍON (PrPsc)
ES08787446T ES2572352T3 (es) 2007-08-23 2008-08-25 Procedimiento de aislamiento y purificación de proteína diana exenta de proteínas priónicas (PrPSC)
CA2696865A CA2696865C (en) 2007-08-23 2008-08-25 A process for isolation and purification of a target protein free of prion protein (prpsc)
DK08787446.7T DK2190486T3 (en) 2007-08-23 2008-08-25 Method for Isolating and Purifying a Target Protein Free of Prion Protein (prpsc)
MX2010001919A MX2010001919A (es) 2007-08-23 2008-08-25 Proceso para aislamiento y purificacion de proteina objetivo libre de proteina prionica.
RU2010110805/10A RU2491292C2 (ru) 2007-08-23 2008-08-25 СПОСОБ ВЫДЕЛЕНИЯ И ОЧИСТКИ ЦЕЛЕВОГО БЕЛКА БЕЗ ПРИМЕСИ ПРИОНОВОГО БЕЛКА PrPsc
EP08787446.7A EP2190486B1 (de) 2007-08-23 2008-08-25 Verfahren zur isolierung und reinigung eines prionenproteinfreien zielproteins (prpsc)
JP2010521447A JP5797404B2 (ja) 2007-08-23 2008-08-25 プリオンタンパク質(PrPsc)を含まない目的タンパク質の単離および精製の方法
AU2008290482A AU2008290482B2 (en) 2007-08-23 2008-08-25 A process for isolation and purification of a target protein free of prion protein (PrPSC)
CN200880104009XA CN101842121B (zh) 2007-08-23 2008-08-25 分离和纯化靶蛋白使其不含朊蛋白(PrPsc)的方法
IL203573A IL203573A (en) 2007-08-23 2010-01-28 Process for Isolation and Purification of Target Protein Without Protein Prpsc
US14/982,148 US20160152661A1 (en) 2007-08-23 2015-12-29 Process for isolation and purification of a target protein free of prion protein (prpsc)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP07114856A EP2027875A1 (de) 2007-08-23 2007-08-23 Verfahren zur Isolierung und Reinigung eines prionenproteinfreien Target-Proteins
EP07114856.3 2007-08-23

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/733,306 A-371-Of-International US9296799B2 (en) 2007-08-23 2008-08-25 Process for isolation and purification of a target protein free of prion protein (PrPSC)
US14/982,148 Division US20160152661A1 (en) 2007-08-23 2015-12-29 Process for isolation and purification of a target protein free of prion protein (prpsc)

Publications (2)

Publication Number Publication Date
WO2009024620A2 true WO2009024620A2 (en) 2009-02-26
WO2009024620A3 WO2009024620A3 (en) 2009-04-23

Family

ID=38720286

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/061068 WO2009024620A2 (en) 2007-08-23 2008-08-25 A process for isolation and purification of a target protein free of prion protein (prpsc)

Country Status (16)

Country Link
US (2) US9296799B2 (de)
EP (3) EP2027875A1 (de)
JP (1) JP5797404B2 (de)
KR (1) KR20100057614A (de)
CN (1) CN101842121B (de)
AU (1) AU2008290482B2 (de)
BR (1) BRPI0815758B1 (de)
CA (1) CA2696865C (de)
DK (2) DK3034097T3 (de)
ES (2) ES2671026T3 (de)
IL (1) IL203573A (de)
MX (1) MX2010001919A (de)
RU (1) RU2491292C2 (de)
TR (1) TR201807044T4 (de)
WO (1) WO2009024620A2 (de)
ZA (1) ZA201001246B (de)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011121020A1 (en) 2010-03-30 2011-10-06 Octapharma Ag A process for purifying vitamin k dependent proteins such as coagulation factor ix
JP2014502259A (ja) * 2010-11-05 2014-01-30 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト ミックスモードクロマトグラフィーによる抗体捕捉の最適化方法
EP2727930A1 (de) * 2011-06-29 2014-05-07 Kyowa Hakko Kirin Co., Ltd. Verfahren zur reinigung von proteinen
US9453045B2 (en) 2010-03-30 2016-09-27 Octapharma Ag Process for the purification of a growth factor protein
US9488625B2 (en) 2010-12-15 2016-11-08 Baxalta GmbH Purification of factor VIII using a conductivity gradient
WO2016207328A1 (en) * 2015-06-24 2016-12-29 Glycotope Gmbh PROCESS FOR THE PURIFICATION OF γ-CARBOXYLATED POLYPEPTIDES
US10188965B2 (en) 2012-12-05 2019-01-29 Csl Behring Gmbh Hydrophobic charge induction chromatographic depletion of a protein from a solution
US10792353B2 (en) 2007-03-14 2020-10-06 Takeda Vaccines, Inc. Virus like particle purification
US11091519B2 (en) 2012-06-22 2021-08-17 Takeda Vaccines, Inc. Purification of virus like particles
US11426680B2 (en) 2012-12-05 2022-08-30 Csl Behring Gmbh Hydrophobic charge induction chromatographic protein depleted solution

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008008872A2 (en) 2006-07-14 2008-01-17 Wisconsin Alumni Research Foundation Adsorptive membranes for trapping viruses
EP2027875A1 (de) * 2007-08-23 2009-02-25 Octapharma AG Verfahren zur Isolierung und Reinigung eines prionenproteinfreien Target-Proteins
US20120183527A1 (en) * 2011-01-18 2012-07-19 Baxter International Inc. Measurement of anti-amyloid antibodies in human blood
AU2012213432B2 (en) * 2011-02-01 2016-10-13 Novo Nordisk A/S Purification of insulin
CN102675414A (zh) * 2011-03-08 2012-09-19 上海天伟生物制药有限公司 糖蛋白中去除/灭活朊病毒的方法
KR102125695B1 (ko) * 2012-10-03 2020-06-24 체에스엘 베링 아게 단백질 정제 방법
CN103336124B (zh) * 2012-12-11 2015-07-15 武汉工业学院 一种检测朊病毒蛋白(PrPSC)的方法和试剂盒
EP3160611A4 (de) * 2014-06-25 2018-02-28 JHL Biotech, Inc. Verfahren und reagenzien zur reinigung von proteinen
DE102016004432A1 (de) * 2016-04-12 2017-10-12 Sartorius Stedim Biotech Gmbh Multimodales Adsorptionsmedium mit multimodalen Liganden, Verfahren zu dessen Herstellung und dessen Verwendung
CN110314414B (zh) * 2019-07-23 2021-10-19 宝锐生物科技泰州有限公司 凝血因子xiii的分离纯化方法及其应用
CN112409476B (zh) * 2020-08-13 2022-03-29 中元汇吉生物技术股份有限公司 四种血液来源蛋白的纯化方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994008686A1 (en) * 1992-10-21 1994-04-28 Cornell Research Foundation, Inc. Multimodal chromatographic separation media and process for usingsame
WO1998000441A1 (en) * 1996-07-01 1998-01-08 Biopure Corporation A method for chromatographic removal of prions
WO2003105911A1 (en) * 2002-06-18 2003-12-24 Common Services Agency Removal of prion infectivity
WO2004024318A1 (en) * 2002-09-13 2004-03-25 Pall Corporation Preparation and use of mixed mode solid substrates for chromatography adsorbents and biochip arrays

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5234991A (en) 1975-07-29 1993-08-10 Pasteur Merieux Serums And Vaccines Porous mineral support coated with an aminated polysaccharide polymer
FR2616437B1 (fr) 1987-06-11 1990-09-07 Ibf Polymeres composites, leur preparation et leur utilisation en chromatographie liquide
US5268097A (en) 1992-06-19 1993-12-07 Sepracor Inc. Passivated and stabilized porous mineral oxide supports and method for the preparation and use of same
AT405119B (de) 1996-06-28 1999-05-25 Poschik Roland Vorrichtung zur betätigung des schiebers eines reissverschlusses, sowie reissverschluss
WO2000043048A1 (en) * 1999-01-19 2000-07-27 Common Services Agency Treating protein-containing liquids
US7750129B2 (en) * 2004-02-27 2010-07-06 Ge Healthcare Bio-Sciences Ab Process for the purification of antibodies
SE0400501D0 (sv) 2004-02-27 2004-02-27 Amersham Biosciences Ab Antibody purification
EP1707634A1 (de) * 2005-03-29 2006-10-04 Octapharma AG Verfahren zur Isolierung von rekombinant hergestellten Proteinen
EP2027875A1 (de) * 2007-08-23 2009-02-25 Octapharma AG Verfahren zur Isolierung und Reinigung eines prionenproteinfreien Target-Proteins
US8329871B2 (en) * 2008-06-24 2012-12-11 Octapharma Ag Process of purifying coagulation factor VIII
WO2011121031A1 (en) * 2010-03-30 2011-10-06 Octapharma Ag Process for the purification of a growth factor protein

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994008686A1 (en) * 1992-10-21 1994-04-28 Cornell Research Foundation, Inc. Multimodal chromatographic separation media and process for usingsame
WO1998000441A1 (en) * 1996-07-01 1998-01-08 Biopure Corporation A method for chromatographic removal of prions
WO2003105911A1 (en) * 2002-06-18 2003-12-24 Common Services Agency Removal of prion infectivity
WO2004024318A1 (en) * 2002-09-13 2004-03-25 Pall Corporation Preparation and use of mixed mode solid substrates for chromatography adsorbents and biochip arrays

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BRIMACOMBE D B ET AL: "Characterization and polyanion-binding properties of purified recombinant prion protein" BIOCHEMICAL JOURNAL, PORTLAND PRESS, LONDON, GB, vol. 342, 1999, pages 605-613, XP002253306 ISSN: 0264-6021 *
BURNOUF ET AL: "Current strategies to prevent transmission of prions by human plasma derivatives" TRANSFUSION CLINIQUE ET BIOLOGIQUE, ARNETTE-BLACKWELL, PARIS, FR, vol. 13, no. 5, 1 February 2007 (2007-02-01), pages 320-328, XP005869901 ISSN: 1246-7820 *
FOSTER P R ET AL: "STUDIES ON THE REMOVAL OF ABNORMAL PRION PROTEIN BY PROCESSES USED IN THE MANUFACTURE OF HUMAN PLASMA PRODUCTS" VOX SANGUINIS, S. KARGER AG, BASEL, CH, vol. 78, no. 2, March 2000 (2000-03), pages 86-95, XP009015791 ISSN: 0042-9007 *
PAN K-M ET AL: "PURIFICATION AND PROPERTIES OF THE CELLULAR PRION PROTEIN FROM SYRIAN HAMSTER BRAIN" PROTEIN SCIENCE, CAMBRIDGE UNIVERSITY PRESS, CAMBRIDGE, GB, vol. 1, 1992, pages 1343-1352, XP002043228 ISSN: 0961-8368 *
THYER J ET AL: "Prion-removal capacity of chromatographic and ethanol precipitation steps used in the production of albumin and immunoglobulins" VOX SANGUINIS, vol. 91, no. 4, 2006, pages 292-300, XP002464418 ISSN: 0042-9007 *
ZEILER B ET AL: "Concentration and removal of prion proteins from biological solutions" BIOTECHNOLOGY AND APPLIED BIOCHEMISTRY, ACADEMIC PRESS, US, vol. 37, no. Pt 2, April 2003 (2003-04), pages 173-182, XP002324177 ISSN: 0885-4513 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10792353B2 (en) 2007-03-14 2020-10-06 Takeda Vaccines, Inc. Virus like particle purification
EP3133157A1 (de) 2010-03-30 2017-02-22 Octapharma AG Verfahren zur reinigung von vitamin-k-abhängigen proteinen
US20130079498A1 (en) * 2010-03-30 2013-03-28 Octapharma Ag Process for purifying vitamin k dependent proteins such as coagulation factor ix
JP2013523689A (ja) * 2010-03-30 2013-06-17 オクタファルマ・アーゲー 第ix凝固因子などのビタミンk依存性タンパク質の精製方法
WO2011121020A1 (en) 2010-03-30 2011-10-06 Octapharma Ag A process for purifying vitamin k dependent proteins such as coagulation factor ix
US9453045B2 (en) 2010-03-30 2016-09-27 Octapharma Ag Process for the purification of a growth factor protein
US10214575B2 (en) 2010-03-30 2019-02-26 Octapharma Ag Process for the purification of a growth factor protein
JP2014502259A (ja) * 2010-11-05 2014-01-30 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト ミックスモードクロマトグラフィーによる抗体捕捉の最適化方法
US9422329B2 (en) 2010-11-05 2016-08-23 Hoffmann-La Roche Inc. Optimized method for antibody capturing by mixed mode chromatography
US9488625B2 (en) 2010-12-15 2016-11-08 Baxalta GmbH Purification of factor VIII using a conductivity gradient
EP2727930A1 (de) * 2011-06-29 2014-05-07 Kyowa Hakko Kirin Co., Ltd. Verfahren zur reinigung von proteinen
US9487566B2 (en) 2011-06-29 2016-11-08 Kyowa Hakko Kirin Co., Ltd Method for purifying protein
EP2727930A4 (de) * 2011-06-29 2014-11-12 Kyowa Hakko Kirin Co Ltd Verfahren zur reinigung von proteinen
US11091519B2 (en) 2012-06-22 2021-08-17 Takeda Vaccines, Inc. Purification of virus like particles
US10188965B2 (en) 2012-12-05 2019-01-29 Csl Behring Gmbh Hydrophobic charge induction chromatographic depletion of a protein from a solution
US11426680B2 (en) 2012-12-05 2022-08-30 Csl Behring Gmbh Hydrophobic charge induction chromatographic protein depleted solution
WO2016207328A1 (en) * 2015-06-24 2016-12-29 Glycotope Gmbh PROCESS FOR THE PURIFICATION OF γ-CARBOXYLATED POLYPEPTIDES

Also Published As

Publication number Publication date
AU2008290482B2 (en) 2013-10-03
BRPI0815758B1 (pt) 2018-08-07
EP2027875A1 (de) 2009-02-25
EP2190486A2 (de) 2010-06-02
IL203573A (en) 2013-11-28
JP5797404B2 (ja) 2015-10-21
CA2696865C (en) 2019-09-17
RU2010110805A (ru) 2011-09-27
ES2572352T3 (es) 2016-05-31
MX2010001919A (es) 2010-03-15
JP2010536832A (ja) 2010-12-02
US20100210821A1 (en) 2010-08-19
EP2190486B1 (de) 2016-03-23
TR201807044T4 (tr) 2018-06-21
CN101842121B (zh) 2013-10-30
CN101842121A (zh) 2010-09-22
DK2190486T3 (en) 2016-07-04
AU2008290482A1 (en) 2009-02-26
CA2696865A1 (en) 2009-02-26
ES2671026T3 (es) 2018-06-04
WO2009024620A3 (en) 2009-04-23
RU2491292C2 (ru) 2013-08-27
DK3034097T3 (en) 2018-06-06
KR20100057614A (ko) 2010-05-31
EP3034097A1 (de) 2016-06-22
EP3034097B1 (de) 2018-02-28
US9296799B2 (en) 2016-03-29
ZA201001246B (en) 2010-10-27
BRPI0815758A2 (pt) 2014-10-21
US20160152661A1 (en) 2016-06-02

Similar Documents

Publication Publication Date Title
EP2190486B1 (de) Verfahren zur isolierung und reinigung eines prionenproteinfreien zielproteins (prpsc)
JP5752558B2 (ja) プロテインaおよびイオン交換クロマトグラフィーによる抗体精製
AU2010277491B2 (en) Method for purifying recombinant ADAMTS13 and other proteins and compositions thereof
Masson et al. A family of serine esterases in lytic granules of cytolytic T lymphocytes
KR101804136B1 (ko) 응고 인자 viii을 정제하는 방법
KR101293504B1 (ko) 혼합형 또는 다중형 수지를 사용하는 인자 vⅰⅰⅰ의 정제
Timmann et al. Two major serum components antigenically related to complement factor H are different glycosylation forms of a single protein with no factor H-like complement regulatory functions.
CA2057926C (en) Chemical products
JP2013525411A (ja) 陰イオン交換樹脂を用いる二価カチオン結合タンパク質の精製方法
CA3085885A1 (en) Protein purification and virus inactivation with alkyl glycosides
AU2013203968B2 (en) A method for polishing albumin
WO2000031121A1 (en) Recombinant factor viii binding peptides
WO2023012828A1 (en) Method to purify an antibody composition using cation exchange chromatography

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880104009.X

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08787446

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 461/DELNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008290482

Country of ref document: AU

Ref document number: 203573

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 20107003554

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2696865

Country of ref document: CA

Ref document number: MX/A/2010/001919

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2010521447

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2008290482

Country of ref document: AU

Date of ref document: 20080825

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2008787446

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2010110805

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 12733306

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0815758

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100223